Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
bioRxiv ; 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39229028

ABSTRACT

Emerging data demonstrate systemic and local inflammation regulate right ventricular (RV) adaption in preclinical and human pulmonary arterial hypertension (PAH). Pathological RV inflammation is targetable as antagonism of glycoprotein-130 (GP130) signaling counteracts pathological microtubule remodeling and improves RV function in rodents. Microtubules control several aspects of cardiomyocyte biology including cellular and nuclear size/structure, t-tubule homeostasis, and the proper localization of connexin-43. The intestinal microbiome regulates systemic inflammation, but the impact of the gut microbiome on the GP130-microtubule axis in RV failure is unknown. Here, we examined how the anti-inflammatory bacteria, Lactobacillus , modulated cellular and physiological RV phenotypes in preclinical and clinical PAH. Lactobacillus supplementation restructured the gut micro/mycobiome, suppressed systemic inflammation, combatted pathological GP130-mediated RV cardiomyocyte microtubule remodeling, and augmented RV function in rodent PAH. Moreover, Lactobacillus was associated with superior RV adaption in human PAH. These data further support the hypothesis that inflammation negatively impacts RV adaption in PAH, and identify the gut microbiome as a potentially targetable regulator of RV function in PAH.

2.
JACC Basic Transl Sci ; 8(3): 239-254, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37034280

ABSTRACT

Intermittent fasting (IF) extends life span via pleotropic mechanisms, but one important molecular mediator is adenosine monophosphate-activated protein kinase (AMPK). AMPK enhances lipid metabolism and modulates microtubule dynamics. Dysregulation of these molecular pathways causes right ventricular (RV) failure in patients with pulmonary arterial hypertension. In rodent pulmonary arterial hypertension, IF activates RV AMPK, which restores mitochondrial and peroxisomal morphology and restructures mitochondrial and peroxisomal lipid metabolism protein regulation. In addition, IF increases electron transport chain protein abundance and activity in the right ventricle. Echocardiographic and hemodynamic measures of RV function are positively associated with fatty acid oxidation and electron transport chain protein levels. IF also combats heightened microtubule density, which normalizes transverse tubule structure.

3.
bioRxiv ; 2023 Dec 02.
Article in English | MEDLINE | ID: mdl-36712076

ABSTRACT

Background: Mitochondrial dysfunction, characterized by impaired lipid metabolism and heightened reactive oxygen species (ROS) generation, results in lipid peroxidation-induced ferroptosis. Ferroptosis is an inflammatory mode of cell death as it both promotes complement activation and recruits macrophages. In pulmonary arterial hypertension (PAH), pulmonary arterial endothelial cells exhibit disrupted lipid metabolism and increased ROS production, and there is ectopic complement deposition and inflammatory macrophage accrual in the surrounding vasculature. However, the integrative effects of ferroptosis on metabolism, cellular landscape changes in the lung, complement induction, and pulmonary vascular remodeling are unknown. Methods: Multi-omics analyses in rodents and a genetic association study in humans evaluated the role of ferroptosis in PAH. Results: Ferrostatin-1, a small-molecule ferroptosis inhibitor, mitigated PAH severity and improved right ventricular function in monocrotaline rats. RNA-seq and proteomics analyses demonstrated ferroptosis was induced with increasingly severe PAH. Metabolomics and proteomics data showed ferroptosis inhibition restructured lung metabolism and altered phosphatidylcholine and phosphatidylethanolamine levels. RNA-seq, proteomics, and confocal microscopy revealed complement activation and pro-inflammatory cytokines/chemokines were suppressed by ferrostatin-1. Additionally, ferrostatin-1 combatted changes in endothelial, smooth muscle, and interstitial macrophage abundances and gene activation patterns in the lungs as revealed by deconvolution RNA-seq. Finally, the presence of six single-nucleotide polymorphisms in ferroptosis genes were independently associated with pulmonary hypertension severity in the Vanderbilt BioVU repository. Conclusions: Rodent and human data nominate ferroptosis as a PAH regulating pathway via its ability to modulate lung lipid metabolism, repress pathogenic complement activation, dampen interstitial macrophage infiltration, and restore the lung cellular environment.

4.
Front Cardiovasc Med ; 9: 940932, 2022.
Article in English | MEDLINE | ID: mdl-36093169

ABSTRACT

Background: Heightened glycolytic flux is associated with right ventricular (RV) dysfunction in pulmonary arterial hypertension (PAH). Methylglyoxal, a glycolysis byproduct, is a highly reactive dicarbonyl that has toxic effects via non-enzymatic post-translational modifications (protein glycation). Methylglyoxal is degraded by the glyoxylase system, which includes the rate-limiting enzyme glyoxylase-1 (GLO1), to combat dicarbonyl stress. However, the potential consequences of excess protein glycation on RV function are unknown. Methods: Bioinformatics analysis of previously identified glycated proteins predicted how protein glycation regulated cardiac biology. Methylglyoxal treatment of H9c2 cardiomyocytes evaluated the consequences of excess protein glycation on mitochondrial respiration. The effects of adeno-associated virus serotype 9-mediated (AAV9) GLO1 expression on RV function in monocrotaline rats were quantified with echocardiography and hemodynamic studies. Immunoblots and immunofluorescence were implemented to probe the effects of AAV-Glo1 on total protein glycation and fatty acid oxidation (FAO) and fatty acid binding protein levels. Results: In silico analyses highlighted multiple mitochondrial metabolic pathways may be affected by protein glycation. Exogenous methylglyoxal minimally altered mitochondrial respiration when cells metabolized glucose, however methylglyoxal depressed FAO. AAV9-Glo1 increased RV cardiomyocyte GLO1 expression, reduced total protein glycation, partially restored mitochondrial density, and decreased lipid accumulation. In addition, AAV9-Glo1 increased RV levels of FABP4, a fatty acid binding protein, and hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunits alpha and beta (HADHA and HADHB), the two subunits of the mitochondrial trifunctional protein for FAO. Finally, AAV9-Glo1 blunted RV fibrosis and improved RV systolic and diastolic function. Conclusion: Excess protein glycation promotes RV dysfunction in preclinical PAH, potentially through suppression of FAO.

SELECTION OF CITATIONS
SEARCH DETAIL